Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dig Dis Sci ; 66(4): 1195-1211, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32415564

RESUMEN

BACKGROUND: Chemopreventive effects of zinc for esophageal cancer have been well documented in animal models. This prospective study explores if a similar, potentially chemopreventive action can be seen in Barrett's esophagus (BE) in humans. AIMS: To determine if molecular evidence can be obtained potentially indicating zinc's chemopreventive action in Barrett's metaplasia. METHODS: Patients with a prior BE diagnosis were placed on oral zinc gluconate (14 days of 26.4 mg zinc BID) or a sodium gluconate placebo, prior to their surveillance endoscopy procedure. Biopsies of Barrett's mucosa were then obtained for miRNA and mRNA microarrays, or protein analyses. RESULTS: Zinc-induced mRNA changes were observed for a large number of transcripts. These included downregulation of transcripts encoding proinflammatory proteins (IL32, IL1ß, IL15, IL7R, IL2R, IL15R, IL3R), upregulation of anti-inflammatory mediators (IL1RA), downregulation of transcripts mediating epithelial-to-mesenchymal transition (EMT) (LIF, MYB, LYN, MTA1, SRC, SNAIL1, and TWIST1), and upregulation of transcripts that oppose EMT (BMP7, MTSS1, TRIB3, GRHL1). miRNA arrays showed significant upregulation of seven miRs with tumor suppressor activity (-125b-5P, -132-3P, -548z, -551a, -504, -518, and -34a-5P). Of proteins analyzed by Western blot, increased expression of the pro-apoptotic protein, BAX, and the tight junctional protein, CLAUDIN-7, along with decreased expression of BCL-2 and VEGF-R2 were noteworthy. CONCLUSIONS: When these mRNA, miRNA, and protein molecular data are considered collectively, a cancer chemopreventive action by zinc in Barrett's metaplasia may be possible for this precancerous esophageal tissue. These results and the extensive prior animal model studies argue for a future prospective clinical trial for this safe, easily-administered, and inexpensive micronutrient, that could determine if a chemopreventive action truly exists.


Asunto(s)
Antineoplásicos/administración & dosificación , Esófago de Barrett/tratamiento farmacológico , Esófago de Barrett/genética , Gluconatos/administración & dosificación , Análisis de Secuencia de ARN/métodos , Administración Oral , Adulto , Anciano , Esófago de Barrett/diagnóstico , Quimioprevención/métodos , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/prevención & control , Femenino , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Proyectos Piloto , Lesiones Precancerosas/diagnóstico , Lesiones Precancerosas/genética , Lesiones Precancerosas/prevención & control , Estudios Prospectivos
2.
Cytogenet Genome Res ; 118(2-4): 196-203, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18000371

RESUMEN

FHIT, at a constitutively active chromosome fragile site, is often a target of chromosomal aberrations and deletion in a large fraction of human tumors. Inactivation of murine Fhit allelessignificantly increases susceptibility of mice to spontaneous and carcinogen-induced tumorigenesis. In this study, transgenic mice, carrying a human FHIT cDNA under control of the endogenous promoter, were produced to determine the effect of Fhit expression, from a nonfragile cDNA transgene outside the fragile region, on carcinogen-induced tumor susceptibility of wildtype and Fhit heterozygous mice. Mice received sufficient oral doses of N-nitrosomethybenzylamine (NMBA) to cause forestomach tumors in >80% of nontransgenic control mice. Although the level of expression of the FHIT transgene in the recombinant mouse strains was much lower than the level of endogenous Fhit expression, the tumor burden in NMBA-treated male transgenic mice was significantly reduced, while female transgenic mice were not protected. To determine if the difference in protection could be due to differences in epigenetic changes at the transgene loci in male versus female mice, we examined expression, hypermethylation and induced re-expression of FHIT transgenes in male and female mice or cells derived from them. The transgene was methylated in male and female mice and in cell lines established from male and female transgenic kidneys, the FHIT locus was both hypermethylated and deacetylated. It is likely that the FHIT transgene is more tightly silenced in female transgenic mice, leading to a lack of protection from tumor induction.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Predisposición Genética a la Enfermedad , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Transgenes , Animales , Secuencia de Bases , Western Blotting , Carcinógenos/toxicidad , Metilación de ADN , Metilasas de Modificación del ADN/antagonistas & inhibidores , Cartilla de ADN , Femenino , Inhibidores de Histona Desacetilasas , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Transgénicos , Neoplasias Experimentales/inducido químicamente , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
3.
J Natl Cancer Inst ; 61(1): 145-50, 1978 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-276623

RESUMEN

Dietary zinc deficiency increases the incidence of and shortens the lag time for induction of esophageal tumors in rats by methylbenzylnitrosamine (MBN). Groups of control and zinc-deficient outbred Charles River CD rats were given 24, 17, 8, or 4 doses of MBN, administered twice weekly by intragastric intubation at doses of 2 mg/kg body weight. Between 58 and 93 days after the beginning of treatment, all rats were killed and examined. The frequency of esophageal tumors in the zinc-deficient groups was significantly higher than in the corresponding control groups. Following ip injection of [methyl-14C]MBN, DNA and RNA of esophagus and liver were more noticeably labeled than kidney, lung, and small intestine, which exhibited only modest labeling. In vitro incubation with [14C]MBN of tissue slices from esophagus, liver, kidney, and small intestine produced similar results. This pattern correlates well with the results of our studies of MBN-induced esophageal tumorigenesis.


Asunto(s)
Carcinoma de Células Escamosas/etiología , Neoplasias Esofágicas/etiología , Nitrosaminas , Zinc/deficiencia , Animales , ADN/metabolismo , Dimetilnitrosamina/análogos & derivados , Esófago/metabolismo , Técnicas In Vitro , Hígado/metabolismo , Masculino , Neoplasias Experimentales/etiología , Nitrosaminas/metabolismo , ARN/metabolismo , Ratas , Factores de Tiempo , Zinc/administración & dosificación
4.
J Natl Cancer Inst ; 93(20): 1525-33, 2001 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-11604475

RESUMEN

BACKGROUND: Nutritional zinc deficiency in rats increases esophageal cell proliferation and the incidence of N-nitrosomethylbenzylamine (NMBA)-induced esophageal tumors. Replenishing zinc with a zinc-sufficient diet reduces these effects in zinc-deficient (ZD) rats. We investigated whether apoptosis was involved in the reduction of NMBA-induced esophageal tumors when ZD rats consumed a zinc-sufficient diet. METHODS: Weanling rats were fed a ZD diet (zinc at 3-4 ppm) for 5 weeks to establish esophageal cell proliferation, then treated once with NMBA (2 mg/kg body weight), and divided into the following five groups (47-100 per group). One ZD group was fed the ZD diet, and four zinc-replenished (ZR) groups, ZR(1), ZR(24), ZR(72), and ZR(432), were fed a zinc-sufficient diet (zinc at 74-75 ppm) beginning 1, 24, 72, and 432 hours, respectively, after NMBA treatment. From 24 hours to 2 weeks after beginning a zinc-sufficient diet, esophagi from all ZR groups were analyzed for apoptosis and cell proliferation; ZD esophagi were the controls. Tumor incidence was determined 15 weeks after zinc replenishment. All statistical tests were two-sided. RESULTS: Zinc replenishment initiated shortly after NMBA treatment effectively reduced esophageal tumorigenesis; 8% (three of 37) of ZR(1), 14% (five of 37) of ZR(24), 19% (five of 26) of ZR(72), and 48% (19 of 40) of ZR(432) rats developed esophageal tumors compared with 93% (14 of 15) of ZD animals (all P<.001). Importantly, 24 and 30 hours after zinc replenishment, esophagi had numerous apoptotic cells (% apoptotic cells: 0 hour = 2.9%, 95% confidence interval [CI] = 2.5% to 3.3%; 24 hours = 9.4%, 95% CI = 8.2% to 10.6%), and the expression of the proapoptotic Bax protein doubled. Within 48 hours, the ZR(1) epithelium was three to five cell layers thick compared with 10-20 layers before zinc replenishment. CONCLUSIONS: Zinc replenishment of NMBA-treated ZD rats rapidly induces apoptosis in esophageal epithelial cells and thereby substantially reduces the development of esophageal cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Dimetilnitrosamina/análogos & derivados , Neoplasias Esofágicas/prevención & control , Papiloma/prevención & control , Zinc/uso terapéutico , Animales , Carbonatos/administración & dosificación , Carcinógenos , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Neoplasias Esofágicas/inducido químicamente , Neoplasias Esofágicas/química , Neoplasias Esofágicas/patología , Esófago/química , Esófago/efectos de los fármacos , Esófago/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hiperplasia , Masculino , Proteínas de Neoplasias/análisis , Papiloma/inducido químicamente , Papiloma/química , Papiloma/patología , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas c-bcl-2/análisis , Ratas , Ratas Sprague-Dawley , Zinc/deficiencia , Zinc/farmacología , Zinc/fisiología , Compuestos de Zinc/administración & dosificación , Proteína X Asociada a bcl-2
5.
J Natl Cancer Inst ; 72(2): 419-25, 1984 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-6582327

RESUMEN

Nine-week-old zinc-sufficient (100 mg zinc/kg feed) and zinc-deficient (7 mg zinc/kg feed) noninbred male Sprague-Dawley rats were given free access 5 days a week to deionized drinking water containing low (0.05%) or high (0.25%) quantities of benzylmethylamine (BMA) and concurrently 0.5% NaNO2. In contrast to the action of the preformed carcinogen N-nitroso-N-benzylmethylamine, which almost invariably produced esophageal tumors, oral administration of its precursors, BMA and NaNO2, resulted in forestomach tumors as well. In both the high- and low-BMA groups given precursors for 16 weeks, the incidence of papillomas in both the esophagus and forestomach was significantly higher in the zinc-deficient than in the zinc-sufficient rats, but zinc deficiency did not significantly increase the yield of forestomach carcinomas. However, when combined high BMA and NaNO2 administration was prolonged to 37 weeks, the yield of forestomach carcinomas was significantly greater in the zinc-deficient than in the zinc-sufficient animals. Because endogenous synthesis of N-nitrosamines from ingested precursors is an important source of human exposure to these carcinogenic compounds and because dietary zinc deficiency might be operating in some areas with a high incidence of esophageal cancer, our data are of more than routine significance.


Asunto(s)
Dimetilnitrosamina/análogos & derivados , Neoplasias Esofágicas/inducido químicamente , Neoplasias Gástricas/inducido químicamente , Zinc/deficiencia , Animales , Bencilaminas/metabolismo , Biotransformación , Dimetilnitrosamina/metabolismo , Neoplasias Esofágicas/patología , Masculino , Nitritos/metabolismo , Ratas , Ratas Endogámicas , Neoplasias Gástricas/patología
6.
Cancer Res ; 58(23): 5380-8, 1998 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-9850069

RESUMEN

Sustained, increased cell proliferation induced by dietary zinc deficiency in rats plays a critical role in esophageal carcinogenesis. It is the determining factor that converts an otherwise nontumorigenic dose of N-nitrosomethylbenzylamine (NMBA) into a highly tumorigenic one. We studied whether the increased esophageal cell proliferation and susceptibility to NMBA-induced carcinogenesis induced by zinc deficiency can be inhibited by alpha-difluoromethylornithine (DFMO), an enzyme-activated, irreversible inhibitor of ornithine decarboxylase (the first enzyme in polyamine synthesis). Weanling rats were divided into four groups: Zn+/DFMO-, Zn+/DFMO+, Zn-/DFMO-, and Zn-/DFMO+. They were fed ad libitum either a zinc-sufficient (Zn+, 75 ppm zinc) or a zinc-deficient (Zn-, 4 ppm zinc) diet and given either deionized water (DFMO-) or 1% DFMO in deionized water (DFMO+). After 5 weeks, 5-19 animals from each group were sacrificed after in vivo 5-bromo-2'-deoxyuridine labeling to detect cells in S phase. The remaining animals in each group were given a single intragastric dose of NMBA at 2 mg/kg and sacrificed 12 weeks later for tumor incidence analysis. At week 5, DFMO treatment greatly decreased (by 48-82%) the levels of putrescine and spermidine in rat esophagus, colon, and liver, irrespective of dietary zinc intake. The increased esophageal cell proliferation induced by dietary zinc deficiency, as measured by the labeling index, the number of labeled cells, and the total number of cells, was substantially reduced by DFMO. This was accompanied by an increase in the rate of apoptosis. In addition, the expression of bax protein, an apoptosis accelerator, was markedly stronger in esophagi from Zn-/DFMO+ animals that showed increased apoptosis, whereas increased expression of bcl-2, an inhibitor of apoptosis, was only seen in the highly proliferative, zinc-deficient esophagus (Zn-/DFMO-). At week 12 after NMBA dosing, DFMO reduced the incidence of esophageal tumors from 80 to 4% in zinc-deficient rats. Our data showed that DFMO effectively inhibited the increased esophageal cell proliferation induced by dietary zinc deficiency and reduced the incidence of esophageal tumors induced by a single dose of NMBA in zinc-deficient animals. Our results also indicate a role for increased apoptosis in the mechanism(s) whereby DFMO brings about the inhibition of cell proliferation and tumor induction. These findings support a role for DFMO as a chemopreventive agent.


Asunto(s)
Anticarcinógenos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinógenos/toxicidad , Cocarcinogénesis , Dimetilnitrosamina/análogos & derivados , Eflornitina/uso terapéutico , Neoplasias Esofágicas/prevención & control , Esófago/efectos de los fármacos , Zinc/deficiencia , Animales , Poliaminas Biogénicas/metabolismo , División Celular/efectos de los fármacos , Dieta , Dimetilnitrosamina/antagonistas & inhibidores , Dimetilnitrosamina/toxicidad , Neoplasias Esofágicas/inducido químicamente , Neoplasias Esofágicas/patología , Esófago/citología , Esófago/metabolismo , Masculino , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas , Ratas Sprague-Dawley , Proteína X Asociada a bcl-2
7.
Cancer Res ; 60(16): 4589-95, 2000 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10969811

RESUMEN

The p16ink4a-cyclin D1/cyclin-dependent kinase 4 (Cdk4)-retinoblastoma (Rb) pathway has emerged as a critical target in oncogenesis. The zinc-deficient (ZD), N-nitrosomethylbenzylamine (NMBA)-induced rat esophageal cancer model provides a tool to study cell proliferation and cell cycle control in cancer initiation. Weanling rats were fed a ZD or zinc-sufficient (ZS) diet for 5 weeks, and then given a dose of NMBA. After 14 weeks, esophageal tumor incidence was 88% in ZD rats with highly proliferative esophagi versus 0% in ZS rats. Expression of p16ink4a, cyclin D1, Cdk4, and Rb in relation to that of proliferating cell nuclear antigen was characterized in esophagi by immunohistochemistry at 0, 24, and 48 h, and 1, 3, 7, 10, and 14 weeks after NMBA treatment. As early as 24 h, proliferating cell nuclear antigen-positive focal hyperplastic lesions were detected in the suprabasal layers of ZD esophagi. At the same time, overexpression of cyclin D1, Cdk4, and Rb was found in the corresponding lesion in adjacent esophageal sections. By contrast, p16ink4a expression was reduced or absent. At all time points, p16ink4a showed reduced nuclear staining in ZD esophagi compared with that in ZS esophagi. In addition, increased expression of the hyperphosphorylated forms of Rb was detected in ZD esophagi by immunoblotting. Importantly, tumors were consistently observed in ZD esophagi at very early time points. These data, obtained using a unique in vivo model for esophageal cancer with rapid tumor induction, provide strong evidence for a link between deregulation of the p16ink4a-cyclin D1/Cdk4-Rb pathway and the initiation of esophageal tumors.


Asunto(s)
Proteínas Portadoras/biosíntesis , Ciclina D1/biosíntesis , Quinasas Ciclina-Dependientes/biosíntesis , Dimetilnitrosamina/análogos & derivados , Neoplasias Esofágicas/patología , Proteínas Proto-Oncogénicas , Proteína de Retinoblastoma/biosíntesis , Transducción de Señal/fisiología , Zinc/deficiencia , Animales , Carcinógenos , División Celular/fisiología , Cocarcinogénesis , Quinasa 4 Dependiente de la Ciclina , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Neoplasias Esofágicas/etiología , Neoplasias Esofágicas/metabolismo , Esófago/citología , Esófago/efectos de los fármacos , Esófago/metabolismo , Fase G1/efectos de los fármacos , Fase G1/fisiología , Inmunohistoquímica , Masculino , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Sprague-Dawley , Fase S/efectos de los fármacos , Fase S/fisiología , Transducción de Señal/efectos de los fármacos
8.
Cancer Res ; 60(11): 2780-5, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10850413

RESUMEN

Hemizygous deletions of the fragile histidine triad (FHIT) gene at human chromosome band 3p14.2 and down-regulation of its gene product are found in the majority of renal cell carcinomas (RCCs). Functional tumor suppressive activity of Fhit in renal cancer cells previously was observed in RCC cell line RC48, which lacks endogenous Fhit expression. To further investigate the potential role of FHIT as a tumor suppressor gene in RCC, we transfected FHIT cDNA expression constructs into RCC cell lines RCC-1 and SN12C, which show low-level expression of endogenous Fhit and reveal an intact von Hippel-Lindau (VHL) gene. Stable transfectants of both cell lines showed no alterations of cell morphology, proliferation kinetics, or cell cycle parameters in vitro. The FHIT gene transfer rate, however, was significantly lower in RCC-1 cells compared with SN12C cells, suggesting a selection against exogenous Fhit expression. In addition, in nude mouse assays, a significant delay of tumor formation was observed for FHIT-transfected RCC-1 cell lines, with outgrowing tumors demonstrating loss of Fhit expression in the majority of cells. In contrast, tumorigenicity of FHIT-transfected SN12C cell clones was not suppressed, despite stable transgene expression. In conclusion, our results demonstrate a selective tumor suppressive activity of Fhit in RCC cells in vivo and suggest that the susceptibility to suppression is not restricted to cancer cells with complete loss of Fhit expression.


Asunto(s)
Ácido Anhídrido Hidrolasas , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Proteínas de Neoplasias , Proteínas/genética , Proteínas/metabolismo , Supresión Genética , Animales , Western Blotting , Ciclo Celular/genética , Cromosomas Humanos Par 3 , ADN Complementario/metabolismo , Regulación hacia Abajo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Transfección , Células Tumorales Cultivadas
9.
Cancer Res ; 58(15): 3401-8, 1998 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-9699672

RESUMEN

The murine Fhit locus maps near the centromere nu proximal Ptprg locus on mouse chromosome 14. The cDNA sequence and structure are similar to those of the human gene, with exons 5-9 encoding the protein. The predominant mRNA in the tissues and cell lines tested was an alternatively spliced form missing exon 3. Most murine cell lines tested, including lines established from normal mouse embryos and tumors, expressed very low or undetectable levels of Fhit mRNA. Most normal mouse tissues expressed wild-type Fhit mRNA, whereas approximately 40% of murine lung carcinomas expressed wild-type and aberrant Fhit RT-PCR products that lacked various exons. Several tumorigenic mouse cell lines exhibited homozygous deletions of Fhit exons. We conclude that the murine Fhit gene, like its human counterpart, is a target of alterations involved in murine carcinogenesis.


Asunto(s)
Ácido Anhídrido Hidrolasas , ADN Complementario/genética , ADN Complementario/aislamiento & purificación , Proteínas de Neoplasias , Biosíntesis de Proteínas , Proteínas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Humanos , Inmunohistoquímica , Ratones , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Transcripción Genética
10.
Physiol Res ; 65(6): 1053-1058, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27539106

RESUMEN

The endothelial barrier function is tightly controlled by a broad range of signaling cascades including nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pathway. It has been proposed that disturbances in NO and cGMP production could interfere with proper endothelial barrier function. In this study, we assessed the effect of interferon-gamma (IFN-gamma), a pro-inflammatory cytokine, on NO and cGMP levels and examined the mechanisms by which NO and cGMP regulate the IFN-gamma-mediated HUVECs hyperpermeability. The flux of fluorescein isothiocyanate-labeled dextran across cell monolayers was used to study the permeability of endothelial cells. Here, we found that IFN-gamma significantly attenuated basal NO concentration and the increased NO levels supplied by a NO donor, sodium nitroprusside (SNP). Besides, application of IFN-gamma also significantly attenuated both the basal cGMP concentration and the increased cGMP production donated by a cell permeable cGMP analogue, 8-bromo-cyclic GMP (8-Br-cGMP). In addition, exposure of the cell monolayer to IFN-gamma significantly increased HUVECs basal permeability. However, L-NAME pretreatment did not suppress IFN-gamma-induced HUVECs hyperpermeability. L-NAME pretreatment followed by SNP or SNP pretreatment partially reduced IFN-gamma-induced HUVECs hyperpermeability. Pretreatment with a guanylate cyclase inhibitor, 6-anilino-5,8-quinolinedione (LY83583), led to a further increase in IFN-gamma-induced HUVECs hyperpermeability. The findings suggest that the mechanism underlying IFN-gamma-induced increased HUVECs permeability is partly related to the inhibition of NO production.


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Interferón gamma/farmacología , Óxido Nítrico/metabolismo , Permeabilidad de la Membrana Celular , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , GMP Cíclico/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Guanilato Ciclasa/antagonistas & inhibidores , Humanos , NG-Nitroarginina Metil Éster/farmacología , Donantes de Óxido Nítrico/farmacología , Nitroprusiato/farmacología
11.
Cancer Epidemiol Biomarkers Prev ; 10(3): 191-9, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11303587

RESUMEN

Alpha-difluoromethylornithine (DFMO) is an irreversible inhibitor of ornithine decarboxylase, the first enzyme in polyamine synthesis. Previous work showed simultaneous administration of DFMO and a zinc-deficient (ZD) diet to weanling rats from the beginning inhibited the onset of zinc-deficiency-induced esophageal cell proliferation by activating apoptosis and reduced the incidence of N-nitrosomethylbenzylamine (NMBA)-induced esophageal cancer. Because esophageal cancer initiation by NMBA is very rapid in ZD rats, this study determined whether DFMO is effective in preventing esophageal carcinogenesis when administered after the establishment of a carcinogenic environment. Weanling rats were given a ZD diet for 5 weeks to establish sustained increased esophageal cell proliferation and then an intragastric dose of NMBA. Thereafter, 20 rats were switched to DFMO-containing water while nine control ZD animals remained on deionized water; all of the animals continued on the ZD diet. Esophagi were collected 15 weeks later. The upper portion was processed for immunohistochemical analysis of cell proliferation, apoptosis, and expression of related genes, and the lower was processed for polyamine content. DFMO substantially reduces the levels of esophageal putrescine and spermidine and esophageal tumor incidence from 89 to 10% in ZD rats. Importantly, DFMO-treated ZD esophagi display increased rate of apoptosis accompanied by intense bax expression and greatly reduced cell proliferation by proliferating cell nuclear antigen expression. In addition, the p16(ink4a)/retinoblastoma control at G1 to S, deregulated in ZD esophagi, is restored after DFMO treatment. These results demonstrate that DFMO, a highly effective chemopreventive agent in esophageal carcinogenesis, reverses and counteracts esophageal cell proliferation/cancer initiation in ZD animals by way of stimulating apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Eflornitina/farmacología , Inhibidores Enzimáticos/farmacología , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/prevención & control , Proteínas Proto-Oncogénicas c-bcl-2 , Análisis de Varianza , Animales , Western Blotting , División Celular/efectos de los fármacos , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/análisis , Dieta , Dimetilnitrosamina/análogos & derivados , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Neoplasias Esofágicas/inducido químicamente , Genes bcl-2 , Inmunohistoquímica , Masculino , Poliaminas/análisis , Antígeno Nuclear de Célula en Proliferación , Proteínas Proto-Oncogénicas/análisis , Ratas , Ratas Sprague-Dawley , Sensibilidad y Especificidad , Zinc/deficiencia , Proteína X Asociada a bcl-2
12.
Cancer Lett ; 143(1): 63-9, 1999 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-10465339

RESUMEN

The effect of zinc deficiency on N-nitrosomethylbenzylamine (NMBA)-induced esophageal tumor formation in rats has been well documented. Our previous work showed that zinc deficiency and its associated increased esophageal cell proliferation were of paramount importance in esophageal tumor development in the NMBA-rat model. However, there has been no report concerning zinc deficiency and NMBA-induced esophageal tumor formation in mice. In this study, weanling C57BL/6 mice were fed ad libitum with either a zinc-sufficient or a zinc-deficient diet containing 3-4 ppm of zinc, and received six intragastric doses of NMBA (2 mg/kg; twice weekly for 3 weeks). The animals were sacrificed 46 weeks later after in vivo bromodeoxyuridine (BrDU) labeling followed by immunohistochemical detection of cells in S-phase. At 46 weeks, the tumor incidences in zinc-deficient mice were 57, 100, and 100% respectively, in the esophagus, forestomach and squamocolumnar junction with the glandular stomach (SCJ), as compared to 17, 39, and 67% in the corresponding tissue of zinc-sufficient mice. The difference between the two dietary groups was significant at P < 0.02 for the esophagus, and P < 0.001 for the forestomach and the SCJ. BrDU labeling revealed that the esophageal labeling index and the number of labeled cells were increased by zinc deficiency. These results support a role of increased cell proliferation in esophageal carcinogenesis in the mouse.


Asunto(s)
Carcinógenos/toxicidad , Dimetilnitrosamina/análogos & derivados , Células Epiteliales/patología , Neoplasias Esofágicas/patología , Esófago/patología , Zinc/deficiencia , Animales , Peso Corporal/efectos de los fármacos , Bromodesoxiuridina/metabolismo , División Celular , Dimetilnitrosamina/toxicidad , Ingestión de Alimentos/efectos de los fármacos , Neoplasias Esofágicas/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Zinc/sangre
13.
Cancer Lett ; 42(3): 217-23, 1988 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-3191474

RESUMEN

The effect of nutritional zinc-deficiency on the activities of O6-alkylguanine:DNA methyltransferase (AGT) in 9 rat tissues including liver, lung, kidney, spleen, brain, esophagus, forestomach, gastric-stomach and small intestine has been examined. Individual tissue extracts prepared from zinc-deficient and pair-fed, zinc-sufficient rats were incubated with N-[3H]methylnitrosourea-methylated calf thymus DNA for 1 h. The activities of AGT in these tissues were measured by two methods: (a) the transfer of the methyl group from O6-methylguanine in substrate DNA to AGT protein, and (b) the determination of the ratio of O6-methylguanine:7-methylguanine remaining in substrate DNA following incubation. AGT activities (expressed as fmol protein methylated/h per mg protein) were significantly reduced in the esophagus, spleen and lungs of zinc-deficient rats as compared to those in their corresponding zinc-sufficient counterparts. The ratio of O6-methylguanine:7-methylguanine was also reduced in the esophagus of the zinc-deficient rat. These results were consistent with our earlier findings that dietary zinc-deficiency enhances nitrosamine-induced esophageal carcinogenesis in rats.


Asunto(s)
Reparación del ADN , Metiltransferasas/metabolismo , Zinc/deficiencia , Animales , Esófago/enzimología , Hígado/enzimología , Masculino , O(6)-Metilguanina-ADN Metiltransferasa , Ratas , Ratas Endogámicas
14.
Cancer Lett ; 22(3): 329-32, 1984 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-6713373

RESUMEN

Administration of N-nitrosodimethylamine (NDMA) to zinc deficient rats intragastrically twice weekly for 8 weeks resulted in the development of forestomach tumors in 63% of treated animals. The tumors were frequently found at the junction between the fore and glandular stomach and were multiple. Smaller numbers of tumors were also present in the more proximal parts of the forestomach. All tumors were squamous papillomas. Similar tumors were not observed in zinc sufficient pair-fed rats given NDMA. These findings showed that NDMA, a non-esophageal and non-forestomach carcinogen in the rat, was tumorigenic in the presence of zinc deficiency.


Asunto(s)
Dimetilnitrosamina/toxicidad , Papiloma/inducido químicamente , Neoplasias Gástricas/inducido químicamente , Zinc/deficiencia , Animales , Esófago/patología , Papiloma/patología , Ratas , Estómago/patología , Neoplasias Gástricas/patología
15.
Cancer Lett ; 30(1): 61-71, 1986 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-3943081

RESUMEN

The influence of dimethylnitrosamine (NDMA), a liver carcinogen and nitrosobenzylmethylamine (NBMA) as esophageal carcinogen on [3H]thymidine incorporation into DNA was studied in the esophagus, liver, forestomach and gastric-stomach of fasted zinc-deficient and pair-fed zinc-sufficient rats, measured 1 h after the thymidine injection. In the untreated animals, dietary zinc deficiency significantly depressed [3H]thymidine incorporation (89%) into the DNA of forestomach only. NDMA, administered 4 h before death at 30 mg/kg, produced 50-55% inhibition in [3H]thymidine incorporation in the esophagus of rats of both dietary groups. This inhibition became more pronounced in the forestomach, reaching 90-94% in the zinc-deficient forestomach and 63-86% in their zinc-sufficient counterparts at NDMA levels ranging from 5 to 20 mg/kg. NBMA at 2 mg/kg produced 60% inhibition in the DNA synthesis of zinc-deficient esophagus and 40% in the corresponding zinc-sufficient ones, this difference being significant at P less than 0.01. On the other hand, [3H]thymidine incorporation in the forestomach DNA was markedly lowered in the presence of NBMA. Recovery of DNA synthesis in the 4 tissues from a single dose of NDMA or NBMA was monitored up to 12 days. Following NDMA injection, [3H]thymidine incorporation in the forestomach of both dietary groups remained inhibited (3% of untreated control) for 5 days, a significant recovery (45% of untreated control) was observed only in the zinc-sufficient animals. Following NBMA injection, [3H]thymidine incorporation was also inhibited in the zinc-deficient esophagus for a longer time than in the zinc-sufficient ones. In autoradiographic studies, the percentage of cells showing 30 or more grains/nucleus was significantly decreased (P less than 0.001) in the NBMA-treated and marginally decreased (P less than 0.05) in the NDMA- or NBMA-treated zinc-deficient and zinc-sufficient rats as compared with the saline-treated zinc-sufficient controls. These results were discussed in the light of our previous findings that NBMA enhanced esophageal tumorigenesis in the zinc-deficient rats and that NDMA, a liver carcinogen produced forestomach tumors in the zinc-deficient but not in the zinc-sufficient rats.


Asunto(s)
ADN/metabolismo , Dimetilnitrosamina/análogos & derivados , Dimetilnitrosamina/toxicidad , Zinc/deficiencia , Animales , Esófago/efectos de los fármacos , Esófago/metabolismo , Mucosa Gástrica/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Especificidad de Órganos , Ratas , Ratas Endogámicas , Estómago/efectos de los fármacos , Timidina/metabolismo
16.
Chem Biol Interact ; 64(1-2): 115-25, 1987.
Artículo en Inglés | MEDLINE | ID: mdl-3121195

RESUMEN

The metabolism of nitrosoproline (NPRO) was re-investigated in uni- and bilaterally nephrectomized rats that have reduced or absent ability to excrete urine. About 1% of the administered radioactivity from L-[U-14C]-NPRO appeared as 14CO2 in the expired air and the production of 14CO2 was time-dependent over a period of 23 h. As compared with sham-operation, uni- or bilateral nephrectomy did not significantly increase the amount of NPRO metabolism, though urinary excretion of radioactivity was decreased in the unilaterally nephrectomized animals. In microsome-mediated and in vitro enzyme-free (Udenfriend-hydroxylating) systems covalent binding of [2,3,4,5-3H]NPRO to exogenous calf thymus DNA was demonstrated. The above findings confirm that in vivo metabolism of NPRO is possible, albeit, to a very small extent.


Asunto(s)
Nitrosaminas/metabolismo , Animales , Dióxido de Carbono/metabolismo , Radioisótopos de Carbono , ADN/metabolismo , Dietilnitrosamina/metabolismo , Masculino , Ratas , Ratas Endogámicas
17.
Mutat Res ; 105(1-2): 29-36, 1982 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6750374

RESUMEN

The mutagenic activity of dimethylnitrosamine activated by rat-liver microsomes in the presence of NADH was compared with that obtained with NADPH. 3 histidine auxotrophic strains of Salmonella underwent reversions after activation with NADH as the sole coenzyme. All 3 tester strains showed a dose-response relationship with dimethylnitrosamine (10-125 mumoles per plate) after NADH-supported activation. With NADH as the sole coenzyme, the most sensitive strain, hisG46, showed a 105-fold increase in mutagenesis frequency as compared with the 230-fold increase obtained with NADPH. Activation of dimethylnitrosamine in the presence of NADH and NADPH, in combination, produced mutagenesis at frequencies above those seen with NADH alone, but less than or equal to those seen with NADPH as the only coenzyme during the activation step. Experiments in vitro showed that microsomal incorporation of carbon from [14C]dimethylnitrosamine was highest in the presence of NADPH, lowest with NADH and reached intermediate levels when both coenzymes were present. The source of the microsomes in all experiments was liver from rats pre-treated with Aroclor 1254.


Asunto(s)
Dimetilnitrosamina/metabolismo , Microsomas Hepáticos/metabolismo , Mutación , Animales , Biotransformación , Dimetilnitrosamina/farmacología , Masculino , Pruebas de Mutagenicidad , NAD/farmacología , Oxidación-Reducción , Ratas , Ratas Endogámicas , Salmonella typhimurium/efectos de los fármacos , Especificidad de la Especie
18.
Pharmacol Biochem Behav ; 26(2): 293-7, 1987 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3575353

RESUMEN

The effects of zinc deficiency on gastric secretion and on cold-restraint stress-induced ulceration in rat stomachs have been studied. Administration of graded zinc deficient diets for 5 weeks significantly depressed the serum zinc concentration and decreased body weight gain in the rats. These diets significantly increased the gastric secretory volume, acid and pepsin. Zinc deficiency produced or aggravated the formation of glandular ulceration in the absence or presence of stress, respectively; it also decreased the mast cell count in the gastric glandular mucosa. It is concluded that zinc deficiency adversely affects the rats by reducing the body weight gain and producing ulceration which is probably mast cell-mediated. On the other hand, it increases gastric secretory functions.


Asunto(s)
Mucosa Gástrica/metabolismo , Úlcera Gástrica/fisiopatología , Estrés Psicológico/fisiopatología , Zinc/deficiencia , Animales , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/patología , Masculino , Mastocitos/citología , Píloro/fisiología , Ratas , Ratas Endogámicas , Úlcera Gástrica/psicología , Zinc/farmacología
19.
Oncogene ; 31(42): 4550-8, 2012 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-22179833

RESUMEN

Chronic inflammation is implicated in the pathogenesis of esophageal squamous cell carcinoma (ESCC). The causes of inflammation in ESCC, however, are undefined. Dietary zinc (Zn)-deficiency (ZD) increases the risk of ESCC. We have previously shown that short-term ZD (6 weeks) in rats induces overexpression of the proinflammatory mediators S100a8 and S100a9 in the esophageal mucosa with accompanying esophageal epithelial hyperplasia. Here we report that prolonged ZD (21 weeks) in rats amplified this inflammation that when combined with non-carcinogenic low doses of the environmental carcinogen, N-nitrosomethylbenzylamine (NMBA) elicited a 66.7% (16/24) incidence of ESCC. With Zn-sufficiency, NMBA produced no cancers (0/21) (P<0.001). At tumor endpoint, the neoplastic ZD esophagus, as compared with Zn-sufficient esophagus, had an inflammatory gene signature with upregulation of numerous cancer-related inflammation genes (CXC and CC chemokines, chemokine receptors, cytokines and Cox-2) in addition to S100a8 and S100a9. This signature was already activated in the earlier dysplastic stage. Additionally, time-course bioinformatics analysis of expression profiles at tumor endpoint and before NMBA exposure revealed that this sustained inflammation was due to ZD rather than carcinogen exposure. Importantly, Zn replenishment reversed this inflammatory signature at both the dysplastic and neoplastic stages of ESCC development, and prevented cancer formation. Thus, the molecular definition of ZD-induced inflammation as a critical factor in ESCC development has important clinical implications with regard to development and prevention of this deadly disease.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Esofágicas/genética , Esófago/metabolismo , Inflamación/genética , Zinc/administración & dosificación , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Calgranulina A/genética , Calgranulina A/metabolismo , Calgranulina B/genética , Calgranulina B/metabolismo , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Análisis por Conglomerados , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Dieta , Dimetilnitrosamina/análogos & derivados , Neoplasias Esofágicas/etiología , Neoplasias Esofágicas/metabolismo , Esófago/efectos de los fármacos , Esófago/patología , Perfilación de la Expresión Génica , Inmunohistoquímica , Inflamación/etiología , Inflamación/metabolismo , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Ratas Sprague-Dawley , Receptores de Quimiocina/genética , Receptores de Quimiocina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Destete , Zinc/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA